the role and regulation of pulmonary artery smooth muscle

10
Review Article The Role and Regulation of Pulmonary Artery Smooth Muscle Cells in Pulmonary Hypertension Shixin He, Tengteng Zhu, and Zhenfei Fang Department of Cardiology, e Second Xiangya Hospital of Central South University, No. 139, Middle Ren-Min Road, Changsha, Hunan 410011, China Correspondence should be addressed to Zhenfei Fang; [email protected] Received 5 December 2019; Accepted 20 July 2020; Published 12 August 2020 Academic Editor: Salvatore Corrao Copyright © 2020 Shixin He et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Pulmonary hypertension (PH) is one of the most devastating cardiovascular diseases worldwide and it draws much attention from numerous scientists. As an indispensable part of pulmonary artery, smooth muscle cells are worthy of being carefully investigated. To elucidate the pathogenesis of PH, several theories focusing on pulmonary artery smooth muscle cells (PASMC), such as hyperproliferation, resistance to apoptosis, and cancer theory, have been proposed and widely studied. Here, we tried to summarize the studies, concentrating on the role of PASMC in the development of PH, feasible molecular basis to intervene, and potential treatment to PH. 1. Introduction Pulmonary hypertension (PH) is a serious global health problem, which is characterized by progressing elevated pulmonary pressures and right heart failure, and mainly affects childbearing women [1]. e mean time from onset of symptoms to diagnosis is about 2 years, the mean survival time of idiopathic/heritable pulmonary arterial hypertension patients from treatment initiation is about 14.7 years, and the 10-year survival rates are 69.5% [2, 3]. Based on recent estimates, in the global population, the prevalence of PH is about 1%, while for individuals aged over 65 years, the number increases to 10%. What is more, about 80% of PH patients are living in developing countries [4]. e feature of PH is intense remodeling of small pul- monary arteries by myofibroblast and smooth muscle cell proliferation, and for familial pulmonary arterial hyper- tension, the bone morphogenetic protein type II receptor (BMPR-II) mutation in pulmonary artery smooth muscle cells contributes to abnormal growth responses to the transforming growth factor (TGF)-beta/bone morphoge- netic protein (BMP) [5]. Compared to previous belief that vasoconstriction acts a vital role in PH pathogenesis [6, 7], there is a tendency to think that excessive proliferation and resistance to apoptosis of PASMC and pulmonary artery endothelial cells (PAEC) are the crucial components of pulmonary vascular remodeling [8]. PASMC has been widely proved to play an important role in the development of various types of pulmonary hypertension. Different mechanisms finally lead to uncontrolled proliferation of PASMC through apoptosis resistance, activated hypoxia- induced factor (HIF), HDAC modification, and inflam- mation, resulting in pulmonary hypertension [9, 10]. According to similar pathophysiological mechanisms, clinical presentation, haemodynamic characteristics, and therapeutic management, the clinical classification of PH is intended to categorize multiple clinical conditions into five groups [11]. Here, we mainly talk about WHO group 1 pulmonary arterial hypertension (PAH). To offer more suitable treatment and precisely evaluate patients’ clinical outcome, the following parameters appear to have the greatest predictive capability: functional class, six-minute walk distance (6MWD), N-terminal pro-brain natriuretic peptide/brain natriuretic peptide (NT-proBNP/BNP) levels, cardiac index, right atrial pressure, and mixed venous ox- ygen saturation (SvO2) [12, 13]. Specific drug treatment of WHO group 1 PAH by targeting the nitric oxide, endothelin, and prostaglandin pathways has been the standard since Hindawi International Journal of Hypertension Volume 2020, Article ID 1478291, 10 pages https://doi.org/10.1155/2020/1478291

Upload: others

Post on 01-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Review ArticleThe Role and Regulation of Pulmonary Artery Smooth MuscleCells in Pulmonary Hypertension

Shixin He, Tengteng Zhu, and Zhenfei Fang

Department of Cardiology,�e Second Xiangya Hospital of Central South University, No. 139, Middle Ren-Min Road, Changsha,Hunan 410011, China

Correspondence should be addressed to Zhenfei Fang; [email protected]

Received 5 December 2019; Accepted 20 July 2020; Published 12 August 2020

Academic Editor: Salvatore Corrao

Copyright © 2020 Shixin He et al. +is is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Pulmonary hypertension (PH) is one of the most devastating cardiovascular diseases worldwide and it draws much attention fromnumerous scientists. As an indispensable part of pulmonary artery, smooth muscle cells are worthy of being carefully investigated.To elucidate the pathogenesis of PH, several theories focusing on pulmonary artery smooth muscle cells (PASMC), such ashyperproliferation, resistance to apoptosis, and cancer theory, have been proposed and widely studied. Here, we tried tosummarize the studies, concentrating on the role of PASMC in the development of PH, feasible molecular basis to intervene, andpotential treatment to PH.

1. Introduction

Pulmonary hypertension (PH) is a serious global healthproblem, which is characterized by progressing elevatedpulmonary pressures and right heart failure, and mainlyaffects childbearing women [1].+emean time from onset ofsymptoms to diagnosis is about 2 years, the mean survivaltime of idiopathic/heritable pulmonary arterial hypertensionpatients from treatment initiation is about 14.7 years, andthe 10-year survival rates are 69.5% [2, 3]. Based on recentestimates, in the global population, the prevalence of PH isabout 1%, while for individuals aged over 65 years, thenumber increases to 10%. What is more, about 80% of PHpatients are living in developing countries [4].

+e feature of PH is intense remodeling of small pul-monary arteries by myofibroblast and smooth muscle cellproliferation, and for familial pulmonary arterial hyper-tension, the bone morphogenetic protein type II receptor(BMPR-II) mutation in pulmonary artery smooth musclecells contributes to abnormal growth responses to thetransforming growth factor (TGF)-beta/bone morphoge-netic protein (BMP) [5]. Compared to previous belief thatvasoconstriction acts a vital role in PH pathogenesis [6, 7],there is a tendency to think that excessive proliferation and

resistance to apoptosis of PASMC and pulmonary arteryendothelial cells (PAEC) are the crucial components ofpulmonary vascular remodeling [8]. PASMC has beenwidely proved to play an important role in the developmentof various types of pulmonary hypertension. Differentmechanisms finally lead to uncontrolled proliferation ofPASMC through apoptosis resistance, activated hypoxia-induced factor (HIF), HDAC modification, and inflam-mation, resulting in pulmonary hypertension [9, 10].

According to similar pathophysiological mechanisms,clinical presentation, haemodynamic characteristics, andtherapeutic management, the clinical classification of PH isintended to categorize multiple clinical conditions into fivegroups [11]. Here, we mainly talk about WHO group 1pulmonary arterial hypertension (PAH). To offer moresuitable treatment and precisely evaluate patients’ clinicaloutcome, the following parameters appear to have thegreatest predictive capability: functional class, six-minutewalk distance (6MWD), N-terminal pro-brain natriureticpeptide/brain natriuretic peptide (NT-proBNP/BNP) levels,cardiac index, right atrial pressure, and mixed venous ox-ygen saturation (SvO2) [12, 13]. Specific drug treatment ofWHO group 1 PAH by targeting the nitric oxide, endothelin,and prostaglandin pathways has been the standard since

HindawiInternational Journal of HypertensionVolume 2020, Article ID 1478291, 10 pageshttps://doi.org/10.1155/2020/1478291

2003. Recently, based on different risk stratification, mon-otherapy or dual-combination therapies, including maci-tentan and sildenafil, riociguat and bosentan, selexipag andendothelin receptor antagonist (ERA) or phosphodiesteraseinhibitor (PDE5i), or both, are recommended [14, 15].

2. Histopathology of Lungs in PH

2.1. Histology of Normal Lung Vessels. +e major role of theright ventricle (RV) is to pump all the blood it receives perbeat into the pulmonary circulation without elevating rightatrial pressure. Normally, blood flow varies with minimumchanges in pulmonary arterial pressure. Although the totalcompliance of the pulmonary circulation is about one-seventh that of the systemic circulation, it stores much lessblood and has the ability to collapse pulmonary vessels aswell as have them distended. +us, the pulmonary circu-lation is able to accommodate increased blood volumeswithout increasing pulmonary artery pressure as much aswould occur on the systemic circulation [16, 17].

2.2. Histopathology of PAHLungVessels. In 1958, Heath andEdwards [18] first described the histologic features of hy-pertensive pulmonary vascular structure changes into sixgrades in patients with congenital septal defects of the heart.+e six grades included retention of fetal type pulmonaryvessels, medial hypertrophy with cellular intimal reaction,progressive fibrous vascular occlusion, progressive gener-alized arterial dilatation with the formation of complexdilatation lesions (plexiform lesions), chronic dilatation withformation of numerous dilatation lesions and pulmonaryhemosiderosis, and necrotizing arteritis. It is widely acceptedthat higher grade is related to worse pulmonary vessels andright heart function. Compared to the control groups, intimaand intima plus media fractional thicknesses of pulmonaryarteries were increased in the PAH group, in accordancewith pulmonary haemodynamic measurements. +ere wereremarkable perivascular inflammation in a mass of PAHlungs and correlated with intima plus media remodeling[19].

Pulmonary vasoconstriction caused by hypoxia wasstudied widely in PH [7]. As a result of global pulmonaryhypoxic vasoconstriction, the right ventricular afterloadcould increase. Chronic hypoxia-induced PH is partly due toinitial pulmonary artery contraction. Pulmonary arterypressures are higher in high-altitude dwellers with chronicmountain sickness, a syndrome including dyspnoea, fatigue,poor sleep, headache, and cyanosis. Hypoxic pulmonaryvascular remodeling also contributes to PH and begins todevelop within the first hours of hypoxic exposure. Hypoxia-induced PH in humans or animals is generally mild ormoderate, but with a substantial afterload on the rightventricle during exercise. In vitro, hypoxia was reported toinhibit myocardial fibre contractility. Pulmonary vascularcontraction plays an important role not only in hypoxic PH,but also in pulmonary arterial hypertension (PAH). Currentpharmacological therapies for PAH mostly target pathwaysregulating endothelial factors with vasoconstrictive/

vasodilatory and have made great achievements in im-proving the exercise capacity, haemodynamics, and time toclinical worsening of PAH patients.

It is increasingly believed that although vasospasm acts arole, pulmonary hypertension is an obstructive lung pan-vasculopathy and different forms of PH present with either apredominance of pulmonary arterial remodeling or veinremodeling or a variable contribution of both [20]. Obvi-ously, there is medial and adventitial thickening of thepulmonary muscular and elastic vessels. +e medial thick-ening is believed to result in hypertrophy and increasedaccumulation of smooth muscle cells as well as increaseddeposition of extracellular matrix proteins, predominantlycollagen and elastin. +e extent of structural changes, in-cluding SMC proliferation, hypertrophy, matrix proteinproduction, and recruitment of adventitial or circulatingcells, in the medial compartment of the pulmonary arterialwall partly determined the severity of chronic hypoxicpulmonary hypertension [21].

3. The Alteration of PASMC in PH

Data from post-mortem studies demonstrated medial hy-pertrophy, PASMC hyperproliferation, and muscle exten-sion into distal arterioles, with important variability betweenindividuals [22–25]. +e accurate regulation of the balancebetween PASMC proliferation and apoptosis is significant inmaintaining the normal integrity of structure and functionin the pulmonary vessels. However, in severe angioproli-ferative PAH, this balance seems to be broken, followingincreased PASMC proliferation and decreased apoptosis,resulting in vessel wall thickening and vascular remodeling[26–31]. Contrast to previous belief that the relationshipbetween pulmonary artery endothelial cells (PAEC) andPASMC is a simple one-way interaction from the endo-thelium to the PASMC, now it is more likely to believe thatmore complicated interactions exist between them [32–34].Under abnormal or irritant conditions, the intricate inter-action of PAEC and PASMC can be altered in the long termso that vascular proliferation and vasocontractility are en-hanced further, which leads to PAH and right heart failure[35–38]. Owing to the characteristics of hyperproliferationand resistance to apoptosis of PASMC in PAH, there is anargument that PAH has something to do with cancer. At themolecular level, PASMC of PAH exhibits many featuressimilar to cancer cells, which gives the chance to explorepotential therapeutic treatments used in cancer to cure PAH[8, 39, 40].

4. Possible Pathways to Act on PASMC

4.1. Role of Ion Channels. It is well known that ions playmany important roles in cell potential, cell contraction, andpH homeostasis, which can influence the proliferation andapoptosis of PASMC. Some studies demonstrated that de-crease of K+ channels affected the PASMC depolarization,then facilitated vascular remodeling, and inhibited PASMCapoptosis. In PAH rat models, restoration of K+ channelsactivity and expression, using dehydroepiandrosterone or

2 International Journal of Hypertension

dichloroacetate, reduced pulmonary vascular remodeling.However, the exact mechanisms by which K+ channels acton PASMC are still controversial [41–51]. Lv et al. foundincreased expression of MicroRNA-206 suppressed potas-sium voltage-gated channel subfamily A member 5 (Kv1.5)and promoted the PASMC proliferation [52].

+e elevated concentration of intracellular Ca2+ wasfound in PAH animal models and patients. +is kind ofphenomenon was not realized through activation of voltage-gated calcium channels (VGCC), but by increase of ca-nonical transient receptor potential (TRPC) proteins, whichinvolved Ca2+-permeable nonselective cation channels(NSCCs). Increased abundance of NSCCs was detected inPAH rat models and patients and inhibition of NSCCs,either pharmacologically or by RNA silencing, effectivelydecreased the concentration of intracellular Ca2+ and pro-liferation of PASMC [53–61]. Song et al. reported thatstromal interaction molecule 2 (STIM2) protein, a Ca2+

sensor in the sarcoplasmic reticulum (SR) membrane, maycontribute to elevated intracellular Ca2+ [62]. What is more,Ca2+ could activate nuclear factor of activated T-cells(NFAT), then suppress K+ channels expression, and lead toPASMC hyperproliferation [63]. It was also proved thathypoxia can cooperate with intracellular Ca2+, which in-creased the expression of aquaporin 1 (AQP1), a membranewater channel, indispensable for PASMC migration. In-creased AQP1 upregulated β-catenin and its target genes(such as c-Myc and cyclin D1), which accelerated theproliferation and migration of PASMC [64–66].

+e normal operation of Na+/H+ exchange (NHE) is es-sential to keep pH homeostasis of PASMC [67, 68]. Studiesshowed that increased expression of NHE isoform 1 (NHE1)can promote the exchange, elevate the pH, and induce theproliferation and migration of PASMC. Although the specificmechanisms are still unclear, it may have something to do withp27 (a cyclin-dependent kinase inhibitor), E2F1 (a nucleartranscription factor), and cytoskeletal re-arrangement [69–75].

4.2. CrucialMolecules. When we talk about PAH, we shouldnever miss hypoxia and hypoxia-inducible factors (HIF).Under the circumstances of hypoxia, increased expressionand decreased degradation result in accumulation of HIF-1α. A lot of studies proved that HIF-1α can influence thePASMC proliferation and mediate pulmonary vascularremodeling, by acting on Ca2+, pH homeostasis, endothelin-1 (ET-1), vascular endothelial growth factor (VEGF), andWarburg effect [76–84].

Endothelin is secreted by endothelial cells and has threeisoforms, among which endothelin-1 (ET-1) is the mostwidely expressed and mediates vascular contraction, cellmigration, and proliferation. In terms to PASMC, ET-1binds to ETA or ETB and then has an impact on decreased K+

channels, elevated intracellular Ca2+, and activation ofNHE1 and Rho kinase (ROCK) signaling, leading to themigration and proliferation of PASMC [85–88].

5-Hydroxytryptamine (5-HT) is well known in de-pression mechanism and it also takes part in the develop-ment of PAH. 5-HT enters PASMC through serotonin

transporter (SERT). +e signaling cascades caused by 5-HTinclude increased reactive oxygen species and activation ofmitogen-activated protein kinase (MAPK) and ROCKpathway, which regulate the expression of genes targetingcell growth and influence PASMC [89–93].

4.3. Important Pathways

4.3.1. Rho Kinase. Rho kinase (ROCK) signaling pathwayplays an indispensable part in vascular contraction andremodeling. Exposed to hypoxia, activation of ROCK inPASMC through Rho B (upstream activators of ROCK)could augment the proliferation and migration of PASMC,resulting in increased pulmonary vascular resistance. +erewere studies stating that long-term use of ROCK inhibitorscould ameliorate vascular remodeling [94–103]. Abe et al.reported that PDGF activated ROCK, suppressed thetranslocation of Smad1 originally induced by bone mor-phogenetic protein 2 (BMP 2), and increased PASMCproliferation [104].

4.3.2. BMP Signaling. Bone morphogenetic protein receptortype 2 (BMPR2) mutations are present in patients withheritable and idiopathic PAH, which reminds us of BMPsignaling’s significant role in the development of PAH. +emutation of BMPR2 could inhibit the antiproliferation effectof BMP2, leading to PAH. BMP can exert its function in away of Smad dependent or independent. BMP/BMPR1interacts with Smad1/5/8, then increasing their binding withSmad4, finally leading to elevated related genes expression.In other ways, BMP activates MAPK, PI3K/AKT, or proteinkinase C (PKC) to influence PASMC. +e impaired controlof BMP signaling may be a common characteristic of PH nomatter what the pathogenesis is [105–110] (Figure 1).

4.3.3. Cancer �eories. As mentioned above, at the molec-ular level, PASMC of PAH exhibits many features similar tocancer cells, making it possible to explore potential thera-peutic treatments used in cancer to cure PAH (reviewed in[40]). Studies showed increase of IL-6, monocyte chemo-tactic protein 1 (MCP-1), and tumor necrosis factor alpha(TNF-α) related to worse clinical outcomes in PAH patients.IL-6 knockout effectively ameliorated PAH in animalmodels. Platelet-derived growth factor (PDGF) mediatedmitogenic signaling and thickening of the pulmonary vascularmedia. +ese growth factors and inflammatory mediatorseventually have an impact on cell growth and survival byMEK/ERK, PI3K/AKT, or JAK/STAT3 pathways. In PASMC,it was reported that activation of STAT3 can upregulate theexpression of proviral integration site for Moloney murineleukemia virus-1 (PIM-1) and then enhance NFAT-mediatedtransactivation, resulting in decreased K+ channels and in-creased intracellular Ca2+. In addition, activation of PI3K/AKT and JAK/STAT3 inhibited the transcription factorForkhead box protein O1 (FOXO1), causing elevated CyclinB1 and D1 and decreased p27, which promoted PASMCproliferation [48, 111–122].

International Journal of Hypertension 3

Mammalian target of rapamycin (mTOR) signaling playsimportant roles in cell metabolism, cell proliferation, andsurvival. Together with other proteins, mTOR forms twoindependent complexes, mTORC1 (mTOR-Raptor) andmTORC2 (mTOR-Rictor). Activation of mTORC1 couldenhance ribosomal protein S6 kinase beta-1 (S6K1) andsuppress eukaryotic translation initiation factor 4E-bindingprotein 1 (4E-BP1), which facilitates cell growth and pro-liferation. On the other hand, mTORC2 is more likely torespond to growth factors, increasing cell survival [123–125].However, Tang et al. reported that mTORC1 and mTORC2had different roles in the development of PAH. Inhibition ofmTORC1 ameliorated pulmonary hypertension, while in-hibition of mTORC2 facilitated spontaneous pulmonaryhypertension and it may result from upregulation of PDGFreceptors in PASMC [126].

+e Hippo signaling pathway is believed to relate tocontrolling organ size. It is constitutive of a cascade of tumorsuppressive kinases mammalian STE20-like protein kinase1/2 (MST1/2) and large tumor suppressor homolog 1/2(LATS1/2), while its downstream molecules include yes-associated protein 1 (YAP) and transcriptional coactivatorwith PDZ-binding motif (TAZ). Inactivation of LATS1/2leads to decrease of YAP and TAZ in cytoplasm and acti-vation of HIF-1α and Notch3 pathways, which plays adeleterious role in the development of PAH [127–133].

Most cancer cells rely on aerobic glycolysis, instead ofdepending on mitochondrial oxidative phosphorylation togenerate energy, a phenomenon termed “the Warburg ef-fect.” +is effect also can be seen in PASMC and PAH.Driven by HIF activation, augmented glycolysis is charac-terized by elevated expression of pivotal proteins in itspathway, such as glucose transporters, hexokinase, pyruvatedehydrogenase kinase (PDK), lactate dehydrogenase (LDH),and 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase3 (PFKFB3). By interacting with PI3K/AKT, ERK1/2, andHIF-1α and altering the morphology and subcellular dis-tribution of mitochondria, Warburg effect increases theproliferation of PASMC in PAH [10, 134–144].

4.3.4. Other Pathways. Peroxisome proliferator-activatedreceptor c (PPARc) regulates mitochondrial gene expressionand biogenesis. Loss of PPARc leads to derangement inmitochondrial structure and function, which has a harmfulimpact on PASMC and PAH [145]. Xie et al. stated thatleptin effectively ameliorated pulmonary vascular remod-eling and PAH, via activation of ERK1/2 and elevated ex-pression of early growth response-1 (Egr-1), resulting in lossof PPARc [146]. In addition, Li et al. reported that activatingprostanoid EP4 receptor (EP4) also decreased the expressionof PPARc through protein kinase A (PKA) pathway andattenuated pulmonary arterial remodeling [147] (Figure 2).

Cyclin-dependent kinases (CDK) are crucial regulatorsof cell cycle and proliferation. Dinaciclib and palbociclibinhibited specific CDK and decreased PASMC proliferationvia cell cycle arrest and interacted with the downstreamCDK-Rb (retinoblastoma protein)-E2F signaling pathway,offering a potential strategy in PAH [148]. Sphingosinekinase 1 (SphK1) is a lipid kinase for phosphorylatingsphingosine to generate sphingosine-1-phosphate (S1P).SphK1/S1P have been reported to relate to cell proliferation,migration, and survival. TGF-β1 could phosphorylateSmad2/3 and then elevate the expression of SphK1 and S1P,which activates Notch3 pathway to promote PASMC pro-liferation [149]. What is more, Sysol et al. reported thatdecreased micro-RNA-1 induced by hypoxia had an effecton the development of PAH via regulation of sphingosinekinase 1 [150].

5. Potential Treatment to PAH

While calcium channel blockers, endothelin receptor an-tagonists, phosphodiesterase type 5 inhibitors and guanylatecyclase stimulators, prostacyclin analogues, and prostacyclinreceptor agonists are the classical specific drug therapies forPAH, their effects still are limited and unsatisfactory. Basedon the molecular pathways mentioned above, tyrosine ki-nase inhibitors (platelet-derived growth factor inhibitors)and serotonin antagonists are being explored, but present

ET-1 Hypoxia 5-HT BMP

ion channels ET A/ET B SERT BMPR

K+ ↓/Ca2+ ↑/NHE(PH↑) HIF↑ ROCK Smad/PI3K/AKT/PKC

Hyperproliferation and resistance to apoptosis of PASMC

Stimulators

Figure 1: Molecular pathways in PASMC (1). ET-1: endothelin-1, 5-HT: serotonin, BMP: bone morphogenetic proteins, ET A/ET Bendothelin receptor A/B, SERT: serotonin transporter, BMPR: bone morphogenetic proteins receptor, NHE: Na+/H+ exchanger, HIF:hypoxia-induced factor, ROCK: Rho kinase, PI3K/AKT: phosphatidylinositide 3-kinase/protein kinase B, PKC: protein kinase C.

4 International Journal of Hypertension

outcomes are not ideal. Moreover, ROCK inhibitors, VEGFreceptor inhibitors, stem cell therapy, mTOR inhibitors,PPAR-c agonist, and strategies aiming at Warburg effect areall in the early phase of research [15, 142–144, 151, 152].

6. Summary

Although the treatment for pulmonary hypertension hasachieved great improvement, it is still not that satisfactory.Owing to its indispensable role in the development ofpulmonary hypertension, PASMC becomes the research hotspot in PH. Further elucidating the molecular basis ofPASMC, including ion channels, HIF, ET-1, ROCK, BMP,PPAR-c, and Warburg effect, could bring hope to PHtreatment.

Conflicts of Interest

+e authors declare that there are no conflicts of interest.

References

[1] V. V. McLaughlin, S. L. Archer, D. B. Badesch et al., “ACCF/AHA 2009 expert consensus document on pulmonary hy-pertension,” Journal of the American College of Cardiology,vol. 53, no. 17, pp. 1573–1619, 2009.

[2] S. Rich, D. R. Dantzker, S. M. Ayres et al., “Primary pul-monary hypertension. A national prospective study,” Annalsof Internal Medicine, vol. 107, no. 2, pp. 216–223, 1987.

[3] A. Ogawa, T. Satoh, Y. Tamura, K. Fukuda, andH. Matsubara, “Survival of Japanese patients with idiopathic/heritable pulmonary arterial hypertension,” AmericanJournal of Cardiology, vol. 119, no. 9, pp. 1479–1484, 2017.

[4] M. M. Hoeper, M. Humbert, R. Souza et al., “A global view ofpulmonary hypertension,” �e Lancet Respiratory Medicine,vol. 4, no. 4, pp. 306–322, 2016.

[5] N. W. Morrell, “Pulmonary hypertension due to BMPR2mutation: a new paradigm for tissue remodeling?” Pro-ceedings of the American �oracic Society, vol. 3, no. 8,pp. 680–686, 2006.

[6] H. L. Motley, A. Cournand, L. Werko, A. Himmelstein, andD. Dresdaleand, “+e influence of short periods of inducedacute anoxia upon pulmonary artery pressures in man,”American Journal of Physiology-Legacy Content, vol. 150,no. 2, pp. 315–320, 1947.

[7] R. Naeije and C. Dedobbeleer, “Pulmonary hypertension andthe right ventricle in hypoxia,” Experimental Physiology,vol. 98, no. 8, pp. 1247–1256, 2013.

[8] O. Boucherat, G. Vitry, I. Trinh, R. Paulin, S. Provencher, andS. Bonnet, “+e cancer theory of pulmonary arterial hy-pertension,” Pulmonary Circulation, vol. 7, no. 2, pp. 285–299, 2017.

[9] V. A. D. J. Perez, “Molecular pathogenesis and currentpathology of pulmonary hypertension,” Heart Failure Re-views, vol. 21, no. 3, pp. 239–257, 2016.

[10] R. Paulin and E. D. Michelakis, “+e metabolic theory ofpulmonary arterial hypertension,” Circulation Research,vol. 115, no. 1, pp. 148–164, 2014.

[11] G. Simonneau, D. Montani, D. S. Celermajer et al., “Hae-modynamic definitions and updated clinical classification of

PDGF

PPAR-γ

PI3K/AKT JAK/STAT3MEK/ERK

MST1/2

IL-6 TNF-α Otherstimulators

Egr-1

Altered mitochondrial structure and function

mTOR FOXO1

4E-BP1 S6K1 Cyclin B1/D1 ↑, P27↓

LATS 1/2

YAP   TAZ

HIF-1α ↑ Notch3

Warburg effect

Hyperproliferation and resistance to apoptosis of PASMC

Figure 2: Molecular pathways in PASMC (2). PDGF: platelet-derived growth factor, IL-6: interleukin-6, TNF-α: tumor necrosis factor-α,PPAR-c: peroxisome proliferator-activated receptor-c, MST 1/2: mammalian sterile 20-like kinases 1/2, Egr-1: early growth response-1,MEK/ERK: mitogen-activated protein kinase/extracellular-signal regulated kinase, PI3K/AKT: phosphatidylinositide 3-kinase/proteinkinase B, JAK/STAT3: Janus kinase/signal transducer and activator of transcription 3, LATS 1/2: large tumor suppressor kinases 1/2, mTOR:mechanistic target of rapamycin, FOXO1: forkhead box protein O1, YAP: yes-associated protein, TAZ: transcriptional coactivator withPDZ-binding motif, 4E-BP1: eukaryotic translation initiation factor 4E-binding protein 1, S6K1: ribosomal protein S6 kinase beta-1, HIF-1α: hypoxia-induced factor-1α.

International Journal of Hypertension 5

pulmonary hypertension,” European Respiratory Journal,vol. 53, no. 1, Article ID 1801913, 2019.

[12] O. Sitbon, R. L. Benza, D. B. Badesch et al., “Validation of twopredictive models for survival in pulmonary arterial hy-pertension,” European Respiratory Journal, vol. 46, no. 1,pp. 152–164, 2015.

[13] A. Boucly, J. Weatherald, M. Humbert, and O. Sitbon, “Riskassessment in pulmonary arterial hypertension,” EuropeanRespiratory Journal, vol. 3, 2018.

[14] D. F. Condon, N. P. Nickel, R. Anderson, S. Mirza, andV. A. D. J. Perez, “+e 6th world symposium on pulmonaryhypertension: what’s old is new,” F1000Research, vol. 8,p. 888, 2019.

[15] N. Galie, M. Humbert, J.-L. Vachiery et al., “2015 ESC/ERSguidelines for the diagnosis and treatment of pulmonaryhypertension: the joint task force for the diagnosis andtreatment of pulmonary hypertension of the European so-ciety of cardiology (ESC) and the European respiratorysociety (ERS): endorsed by: association for European pae-diatric and congenital cardiology (AEPC), internationalsociety for heart and lung transplantation (ISHLT),” Euro-pean Heart Journal, vol. 37, no. 1, pp. 67–119, 2016.

[16] M. R. Pinsky, “+e right ventricle: interaction with thepulmonary circulation,” Critical Care, vol. 20, no. 1, p. 266,2016.

[17] Y.-Y. He, C.-L. Liu, X. Li, R.-J. Li, L.-L. Wang, and K.-L. He,“Salubrinal attenuates right ventricular hypertrophy anddysfunction in hypoxic pulmonary hypertension of rats,”Vascular Pharmacology, vol. 87, pp. 190–198, 2016.

[18] D. Heath and J. E. Edwards, “+e pathology of hypertensivepulmonary vascular disease; a description of six grades ofstructural changes in the pulmonary arteries with specialreference to congenital cardiac septal defects,” Circulation,vol. 18, no. 4, pp. 533–547, 1958.

[19] E. Stacher, B. B. Graham, J. M. Hunt et al., “Modern agepathology of pulmonary arterial hypertension,” AmericanJournal of Respiratory and Critical Care Medicine, vol. 186,no. 3, pp. 261–272, 2012.

[20] R. M. Tuder, S. L. Archer, P. Dorfmuller et al., “Relevantissues in the pathology and pathobiology of pulmonaryhypertension,” Journal of the American College of Cardiology,vol. 62, no. 25, pp. D4–D12, 2013.

[21] K. R. Stenmark, K. A. Fagan, and M. G. Frid, “Hypoxia-induced pulmonary vascular remodeling: cellular and mo-lecular mechanisms,” Circulation Research, vol. 99, no. 7,pp. 675–691, 2006.

[22] R. L. Naeye, “Hypoxemia and pulmonary hypertension. Astudy of the pulmonary vasculature,” Archives of Pathology,vol. 71, pp. 447–452, 1961.

[23] R. L. Naeye, “Children at high altitude: pulmonary and renalabnormalities,” Circulation Research, vol. 16, no. 1,pp. 33–38, 1965.

[24] J. Arias-Stella and M. Saldana, “+e terminal portion of thepulmonary arterial tree in people native to high altitudes,”Circulation, vol. 28, no. 5, pp. 915–925, 1963.

[25] D. Heath, P. Smith, D. J. Rios, D. Williams, and P. Harris,“Small pulmonary arteries in some natives of La Paz, Boli-via,” �orax, vol. 36, no. 8, pp. 599–604, 1981.

[26] M. Rabinovitch, “Elastase and the pathobiology of unex-plained pulmonary hypertension,” Chest, vol. 114, no. 3,pp. 213S–224S, 1998.

[27] L. J. Rubin, “Cellular and molecular mechanisms responsiblefor the pathogenesis of primary pulmonary hypertension,”Pediatric Pulmonology, vol. 27, no. S18, pp. 194–197, 1999.

[28] C. A. Wagenvoort, “+e pathology of primary pulmonaryhypertension,” Journal of Pathology, vol. 4, 1970.

[29] J. D. Wohrley, M. G. Frid, E. P. Moiseeva, E. C. Orton,J. K. Belknap, and K. R. Stenmark, “Hypoxia selectivelyinduces proliferation in a specific subpopulation of smoothmuscle cells in the bovine neonatal pulmonary arterialmedia,” Journal of Clinical Investigation, vol. 96, no. 1,pp. 273–281, 1995.

[30] J. Diez, M. A. Fortuno, G. Zalba et al., “Altered regulation ofsmooth muscle cell proliferation and apoptosis in smallarteries of spontaneously hypertensive rats,” European HeartJournal, vol. 19, pp. G29–G33, 1998.

[31] S. Sakao, K. Tatsumi, and N. F. Voelkel, “Endothelial cellsand pulmonary arterial hypertension: apoptosis, prolifera-tion, interaction and transdifferentiation,” Respiratory Re-search, vol. 10, no. 1, 2009.

[32] M. Balcells, J. Martorell, C. Olive et al., “Smooth muscle cellsorchestrate the endothelial cell response to flow and injury,”Circulation, vol. 121, no. 20, pp. 2192–2199, 2010.

[33] M. Billaud, A. W. Lohman, S. R. Johnstone, L. A. Biwer,S. Mutchler, and B. E. Isakson, “Regulation of cellularcommunication by signaling microdomains in the bloodvessel wall,” Pharmacological Reviews, vol. 66, no. 2,pp. 513–569, 2014.

[34] R. Nogueira-Ferreira, R. Ferreira, and T. Henriques-Coelho,“Cellular interplay in pulmonary arterial hypertension:implications for new therapies,” Biochimica et BiophysicaActa (BBA)-Molecular Cell Research, vol. 1843, no. 5,pp. 885–893, 2014.

[35] D. D. Ivy, S. H. Abman, R. J. Barst et al., “Pediatric pul-monary hypertension,” Journal of the American College ofCardiology, vol. 62, no. 25, pp. D117–D126, 2013.

[36] E. D. Austin, S. M. Kawut, M. T. Gladwin, and S. H. Abman,“Pulmonary hypertension: NHLBI workshop on the primaryprevention of chronic lung diseases,” Annals of the American�oracic Society, vol. 11, no. 3, pp. S178–S185, 2014.

[37] C. Guignabert, L. Tu, B. Girerd et al., “New molecular targetsof pulmonary vascular remodeling in pulmonary arterialhypertension: importance of endothelial communication,”Chest, vol. 147, no. 2, pp. 529–537, 2015.

[38] Y. Gao, T. Chen, and J. U. Raj, “Endothelial and smoothmuscle cell interactions in the pathobiology of pulmonaryhypertension,” American Journal of Respiratory Cell andMolecular Biology, vol. 54, no. 4, pp. 451–460, 2016.

[39] N. F. Voelkel, C. Cool, S. D. Lee, L. Wright, M. W. Geraci,and R. M. Tuder, “Primary pulmonary hypertension betweeninflammation and cancer,” Chest, vol. 114, no. 3,pp. 225S–230S, 1998.

[40] C. Guignabert, L. Tu, M. L. Hiress et al., “Pathogenesis ofpulmonary arterial hypertension: lessons from cancer,”European Respiratory Review, vol. 22, no. 130, pp. 543–551,2013.

[41] H. Suzuki and B. M. Twarog, “Membrane properties ofsmooth muscle cells in pulmonary hypertensive rats,”American Journal of Physiology-Heart and CirculatoryPhysiology, vol. 242, no. 5, pp. H907–H915, 1982.

[42] L. A. Shimoda and J. Polak, “Hypoxia. 4. Hypoxia and ionchannel function,” American Journal of Physiology-CellPhysiology, vol. 300, no. 5, pp. C951–C967, 2011.

[43] J. X. Yuan, A. M. Aldinger, M. Juhaszova et al., “Dysfunc-tional voltage-gated K+ channels in pulmonary arterysmooth muscle cells of patients with primary pulmonaryhypertension,” Circulation, vol. 98, no. 14, pp. 1400–1406,1998.

6 International Journal of Hypertension

[44] E. D. Burg, C. V. Remillard, and J. X.-J. Yuan, “Potassiumchannels in the regulation of pulmonary artery smoothmuscle cell proliferation and apoptosis: pharmacother-apeutic implications,” British Journal of Pharmacology,vol. 153, no. 1, pp. S99–S111, 2008.

[45] E. D. Michelakis, M. S. McMurtry, X.-C. Wu et al.,“Dichloroacetate, a metabolic modulator, prevents and re-verses chronic hypoxic pulmonary hypertension in rats: role ofincreased expression and activity of voltage-gated potassiumchannels,” Circulation, vol. 105, no. 2, pp. 244–250, 2002.

[46] S. Bonnet, E. D. Michelakis, C. J. Porter et al., “An abnormalmitochondrial-hypoxia inducible factor-1α-Kv channelpathway disrupts oxygen sensing and triggers pulmonaryarterial hypertension in fawn hooded rats: similarities tohuman pulmonary arterial hypertension,” Circulation,vol. 113, no. 22, pp. 2630–2641, 2006.

[47] X.-J. Yuan, J. Wang, M. Juhaszova, S. P. Gaine, andL. J. Rubin, “Attenuated K+ channel gene transcription inprimary pulmonary hypertension,” �e Lancet, vol. 351,no. 9104, pp. 726-727, 1998.

[48] S. Bonnet, G. Rochefort, G. Sutendra et al., “+e nuclearfactor of activated Tcells in pulmonary arterial hypertensioncan be therapeutically targeted,” Proceedings of the NationalAcademy of Sciences, vol. 104, no. 27, pp. 11418–11423, 2007.

[49] Z. I. Pozeg, E. D. Michelakis, M. S. McMurtry et al., “In vivogene transfer of the O2-sensitive potassium channel Kv1.5reduces pulmonary hypertension and restores hypoxicpulmonary vasoconstriction in chronically hypoxic rats,”Circulation, vol. 107, no. 15, pp. 2037–2044, 2003.

[50] S. Bonnet, E. Dumas-De-La-Roque, H. Begueret et al.,“Dehydroepiandrosterone (DHEA) prevents and reverseschronic hypoxic pulmonary hypertension,” Proceedings ofthe National Academy of Sciences, vol. 100, no. 16,pp. 9488–9493, 2003.

[51] M. S. McMurtry, S. Bonnet, X. Wu et al., “Dichloroacetateprevents and reverses pulmonary hypertension by inducingpulmonary artery smooth muscle cell apoptosis,” CirculationResearch, vol. 95, no. 8, pp. 830–840, 2004.

[52] Y. Lv, L. Fu, Z. Zhang et al., “Increased expression ofMicroRNA-206 inhibits potassium voltage-gated channelsubfamily A member 5 in pulmonary arterial smooth musclecells and is related to exaggerated pulmonary artery hy-pertension following intrauterine growth retardation inrats,” Journal of the American Heart Association, vol. 8, no. 2,Article ID e10456, 2019.

[53] J. Wang, L. Weigand, W. Lu, J. T. Sylvester, G. L. Semenza,and L. A. Shimoda, “Hypoxia inducible factor 1 mediateshypoxia-induced TRPC expression and elevated intracellularCa2+ in pulmonary arterial smooth muscle cells,” CirculationResearch, vol. 98, no. 12, pp. 1528–1537, 2006.

[54] M.-J. Lin, G. P. H. Leung, W.-M. Zhang et al., “Chronichypoxia-induced upregulation of store-operated and re-ceptor-operated Ca2+ channels in pulmonary arterial smoothmuscle cells: a novel mechanism of hypoxic pulmonaryhypertension,” Circulation Research, vol. 95, no. 5,pp. 496–505, 2004.

[55] V. A. Golovina, O. Platoshyn, C. L. Bailey et al., “UpregulatedTRP and enhanced capacitative Ca2+ entry in human pul-monary artery myocytes during proliferation,” AmericanJournal of Physiology-Heart and Circulatory Physiology,vol. 280, no. 2, pp. H746–H755, 2001.

[56] J. W. Landsberg and J. X.-J. Yuan, “Calcium and TRPchannels in pulmonary vascular smooth muscle cell prolif-eration,” Physiology, vol. 19, no. 2, pp. 44–50, 2004.

[57] K. Leggett, J. Maylor, C. Undem et al., “Hypoxia-inducedmigration in pulmonary arterial smooth muscle cells re-quires calcium-dependent upregulation of aquaporin 1,”American Journal of Physiology-Lung Cellular and MolecularPhysiology, vol. 303, no. 4, pp. L343–L353, 2012.

[58] X.-R. Liu, M.-F. Zhang, N. Yang et al., “Enhanced store-op-erated Ca2+ entry and TRPC channel expression in pulmonaryarteries of monocrotaline-induced pulmonary hypertensiverats,” American Journal of Physiology-Cell Physiology, vol. 302,no. 1, pp. C77–C87, 2012.

[59] M. Y. Song, A. Makino, and J. X.-J. Yuan, “STIM2 con-tributes to enhanced store-operated Ca entry in pulmonaryartery smooth muscle cells from patients with idiopathicpulmonary arterial hypertension,” Pulmonary Circulation,vol. 1, no. 1, pp. 84–94, 2011.

[60] Y. Yu, I. Fantozzi, C. V. Remillard et al., “Enhanced ex-pression of transient receptor potential channels in idio-pathic pulmonary arterial hypertension,” Proceedings of theNational Academy of Sciences, vol. 101, no. 38, pp. 13861–13866, 2004.

[61] N. Kunichika, J. W. Landsberg, Y. Yu et al., “Bosentan in-hibits transient receptor potential channel expression inpulmonary vascular myocytes,” American Journal of Respi-ratory and Critical Care Medicine, vol. 170, no. 10,pp. 1101–1107, 2004.

[62] S. Song, S. G. Carr, K. M. McDermott et al., “STIM2 (stromalinteraction molecule 2)-mediated increase in resting cyto-solic free Ca2+ concentration stimulates PASMC prolifera-tion in pulmonary arterial hypertension,” Hypertension,vol. 71, no. 3, pp. 518–529, 2018.

[63] R.-L. He, Z.-J. Wu, X.-R. Liu, L.-X. Gui, R.-X. Wang, andM.-J. Lin, “Calcineurin/NFAT signaling modulates pulmo-nary artery smooth muscle cell proliferation, migration andapoptosis in monocrotaline-induced pulmonary arterialhypertension rats,” Cellular Physiology and Biochemistry,vol. 49, no. 1, pp. 172–189, 2018.

[64] S. Saadoun, M. C. Papadopoulos, M. Hara-Chikuma, andA. S. Verkman, “Impairment of angiogenesis and cell mi-gration by targeted aquaporin-1 gene disruption,” Nature,vol. 434, no. 7034, pp. 786–792, 2005.

[65] E. Monzani, R. Bazzotti, C. Perego, and C. A. M. L. Porta,“AQP1 is not only a water channel: it contributes to cellmigration through Lin7/beta-catenin,” PLoS One, vol. 4,no. 7, Article ID e6167, 2009.

[66] X. Yun, H. Jiang, N. Lai, J. Wang, and L. A. Shimoda,“Aquaporin 1-mediated changes in pulmonary arterialsmooth muscle cell migration and proliferation involve beta-catenin,” American Journal of Physiology-Lung Cellular andMolecular Physiology, vol. 313, no. 5, pp. L889–L898, 2017.

[67] J. A. Madden, D. E. Ray, P. A. Keller, and J. G. Kleinman,“Ion exchange activity in pulmonary artery smooth musclecells: the response to hypoxia,” American Journal of Physi-ology-Lung Cellular andMolecular Physiology, vol. 280, no. 2,pp. L264–L271, 2001.

[68] D. A. Quinn, T. W. Honeyman, P. M. Joseph,B. T.+ompson, C. A. Hales, and C. R. Scheid, “Contributionof Na+/H+ exchange to pH regulation in pulmonary arterysmooth muscle cells,” American Journal of Respiratory Celland Molecular Biology, vol. 5, no. 6, pp. 586–591, 1991.

[69] D. A. Quinn, C. G. Dahlberg, J. P. Bonventre et al., “+e roleof Na+/H+ exchange and growth factors in pulmonary arterysmooth muscle cell proliferation,” American Journal of Re-spiratory Cell and Molecular Biology, vol. 14, no. 2,pp. 139–145, 1996.

International Journal of Hypertension 7

[70] E. J. Rios, M. Fallon, J. Wang, and L. A. Shimoda, “Chronichypoxia elevates intracellular pH and activates Na+/H+ ex-change in pulmonary arterial smooth muscle cells,” Amer-ican Journal of Physiology-Lung Cellular and MolecularPhysiology, vol. 289, no. 5, pp. L867–L874, 2005.

[71] L. A. Shimoda, M. Fallon, S. Pisarcik, J. Wang, andG. L. Semenza, “HIF-1 regulates hypoxic induction of NHE1expression and alkalinization of intracellular pH in pul-monary arterial myocytes,” American Journal of Physiology-Lung Cellular and Molecular Physiology, vol. 291, no. 5,pp. L941–L949, 2006.

[72] D. A. Quinn, H.-K. Du, B. T. +ompson, and C. A. Hales,“Amiloride analogs inhibit chronic hypoxic pulmonaryhypertension,” American Journal of Respiratory and CriticalCare Medicine, vol. 157, no. 4, pp. 1263–1268, 1998.

[73] L. Yu, D. A. Quinn, H. G. Garg, and C. A. Hales, “Deficiencyof the NHE1 gene prevents hypoxia-induced pulmonaryhypertension and vascular remodeling,” American Journal ofRespiratory and Critical Care Medicine, vol. 177, no. 11,pp. 1276–1284, 2008.

[74] L. Yu and C. A. Hales, “Silencing of sodium-hydrogen ex-changer 1 attenuates the proliferation, hypertrophy, andmigration of pulmonary artery smooth muscle cells viaE2F1,” American Journal of Respiratory Cell and MolecularBiology, vol. 45, no. 5, pp. 923–930, 2011.

[75] S. P. Denker, D. C. Huang, J. Orlowski, H. Furthmayr, andD. L. Barber, “Direct binding of the Na-H exchanger NHE1to ERM proteins regulates the cortical cytoskeleton and cellshape independently of H+ translocation,” Molecular Cell,vol. 6, no. 6, pp. 1425–1436, 2000.

[76] G. L. Semenza, “Pulmonary vascular responses to chronichypoxia mediated by hypoxia-inducible factor 1,” Proceed-ings of the American�oracic Society, vol. 2, no. 1, pp. 68–70,2005.

[77] N. R. Prabhakar and G. L. Semenza, “Adaptive and mal-adaptive cardiorespiratory responses to continuous andintermittent hypoxia mediated by hypoxia-inducible factors1 and 2,” Physiological Reviews, vol. 92, no. 3, pp. 967–1003,2012.

[78] A. Y. Yu, L. A. Shimoda, N. V. Iyer et al., “Impairedphysiological responses to chronic hypoxia in mice partiallydeficient for hypoxia-inducible factor 1alpha,” Journal ofClinical Investigation, vol. 103, no. 5, pp. 691–696, 1999.

[79] L. A. Shimoda and G. L. Semenza, “HIF and the lung: role ofhypoxia-inducible factors in pulmonary development anddisease,” American Journal of Respiratory and Critical CareMedicine, vol. 183, no. 2, pp. 152–156, 2011.

[80] L. A. Shimoda, “55th Bowditch Lecture: effects of chronichypoxia on the pulmonary circulation: role of HIF-1,”Journal of Applied Physiology, vol. 113, no. 9, pp. 1343–1352,2012.

[81] N. V. Iyer, L. E. Kotch, F. Agani et al., “Cellular and de-velopmental control of O2 homeostasis by hypoxia-induciblefactor 1 alpha,” Genes & Development, vol. 12, no. 2,pp. 149–162, 1998.

[82] L. A. Shimoda, “Hypoxic regulation of ion channels andtransporters in pulmonary vascular smooth muscle,” Ad-vances in Experimental Medicine and Biology, vol. 661,pp. 221–235, 2010.

[83] C.-C. Wang, L. Ying, E. A. Barnes et al., “Pulmonary arterysmooth muscle cell HIF-1alpha regulates endothelin ex-pression via microRNA-543,” American Journal of Physiol-ogy-Lung Cellular and Molecular Physiology, vol. 315, no. 3,pp. L422–L431, 2018.

[84] X. He, S. Song, R. J. Ayon et al., “Hypoxia selectivelyupregulates cation channels and increases cytosolic [Ca2+] inpulmonary, but not coronary, arterial smooth muscle cells,”American Journal of Physiology-Cell Physiology, vol. 314,no. 4, pp. C504–C517, 2018.

[85] D. Shao, J. E. S. Park, and S. J. Wort, “+e role of endothelin-1 in the pathogenesis of pulmonary arterial hypertension,”Pharmacological Research, vol. 63, no. 6, pp. 504–511, 2011.

[86] L. A. Shimoda, J. S. K. Sham, Q. Liu, and J. T. Sylvester,“Acute and chronic hypoxic pulmonary vasoconstriction: acentral role for endothelin-1?” Respiratory Physiology &Neurobiology, vol. 132, no. 1, pp. 93–106, 2002.

[87] E. M. Whitman, S. Pisarcik, T. Luke et al., “Endothelin-1mediates hypoxia-induced inhibition of voltage-gated K+

channel expression in pulmonary arterial myocytes,”American Journal of Physiology-Lung Cellular and MolecularPhysiology, vol. 294, no. 2, pp. L309–L318, 2008.

[88] J. C. Huetsch, J. Walker, C. Undem et al., “Rho kinase andNa+/H+ exchanger mediate endothelin-1-induced pulmonaryarterial smooth muscle cell proliferation and migration,”Physiological Reports, vol. 6, no. 9, Article ID e13698, 2018.

[89] M. R. MacLean, D. Alexander, A. Stirrat et al., “Contractileresponses to human urotensin-II in rat and human pul-monary arteries: effect of endothelial factors and chronichypoxia in the rat,” British Journal of Pharmacology, vol. 130,no. 2, pp. 201–204, 2000.

[90] S. Eddahibi, V. Fabre, C. Boni et al., “Induction of serotonintransporter by hypoxia in pulmonary vascular smooth musclecells. Relationship with the mitogenic action of serotonin,”Circulation Research, vol. 84, no. 3, pp. 329–336, 1999.

[91] K. M. Mair, M. R. MacLean, I. Morecroft, Y. Dempsie, andT. M. Palmer, “Novel interactions between the 5-HTtransporter, 5-HT1B receptors and Rho kinase in vivo and inpulmonary fibroblasts,” British Journal of Pharmacology,vol. 155, no. 4, pp. 606–616, 2008.

[92] M. R. Maclean and Y. Dempsie, “+e serotonin hypothesis ofpulmonary hypertension revisited,” Advances in Experi-mental Medicine and Biology, vol. 661, pp. 309–322, 2010.

[93] M. R. MacLean, “+e serotonin hypothesis in pulmonaryhypertension revisited: targets for novel therapies (2017Grover Conference Series),” Pulmonary Circulation, vol. 8,no. 2, Article ID 204589401875912, 2018.

[94] M. Oka, N. Homma, L. Taraseviciene-Stewart et al., “Rhokinase-mediated vasoconstriction is important in severeocclusive pulmonary arterial hypertension in rats,” Circu-lation Research, vol. 100, no. 6, pp. 923–929, 2007.

[95] J. P. Ward and I. F. McMurtry, “Mechanisms of hypoxicpulmonary vasoconstriction and their roles in pulmonaryhypertension: new findings for an old problem,” CurrentOpinion in Pharmacology, vol. 9, no. 3, pp. 287–296, 2009.

[96] A. L. Firth, I. W. Choi, and W. S. Park, “Animal models ofpulmonary hypertension: rho kinase inhibition,” Progress inBiophysics and Molecular Biology, vol. 109, no. 3, pp. 67–75,2012.

[97] M. Oka, K. A. Fagan, P. L. Jones, and I. F. McMurtry,“+erapeutic potential of RhoA/Rho kinase inhibitors inpulmonary hypertension,” British Journal of Pharmacology,vol. 155, no. 4, pp. 444–454, 2008.

[98] X. Yang, P. J. Lee, L. Long, R. C. Trembath, andN. W. Morrell, “BMP4 induces HO-1 via a Smad-inde-pendent, p38MAPK-dependent pathway in pulmonary ar-tery myocytes,” American Journal of Respiratory Cell andMolecular Biology, vol. 37, no. 5, pp. 598–605, 2007.

8 International Journal of Hypertension

[99] W. T. Gerthoffer, “Mechanisms of vascular smooth musclecell migration,” Circulation Research, vol. 100, no. 5,pp. 607–621, 2007.

[100] Y. Liu, Y. J. Suzuki, R. M. Day, and B. L. Fanburg, “Rhokinase-induced nuclear translocation of ERK1/ERK2 insmooth muscle cell mitogenesis caused by serotonin,” Cir-culation Research, vol. 95, no. 6, pp. 579–586, 2004.

[101] Y. Fukumoto, T. Matoba, A. Ito et al., “Acute vasodilatoreffects of a Rho-kinase inhibitor, fasudil, in patients withsevere pulmonary hypertension,” Heart, vol. 91, no. 3,pp. 391-392, 2005.

[102] K. Ishikura, N. Yamada, M. Ito et al., “Beneficial acute effectsof rho-kinase inhibitor in patients with pulmonary arterialhypertension,” Circulation Journal, vol. 70, no. 2, pp. 174–178, 2006.

[103] K. A. Fagan, M. Oka, N. R. Bauer et al., “Attenuation of acutehypoxic pulmonary vasoconstriction and hypoxic pulmo-nary hypertension in mice by inhibition of Rho-kinase,”American Journal of Physiology-Lung Cellular and MolecularPhysiology, vol. 287, no. 4, pp. L656–L664, 2004.

[104] K. Abe, H. Shimokawa, K. Morikawa et al., “Long-termtreatment with a Rho-kinase inhibitor improves mono-crotaline-induced fatal pulmonary hypertension in rats,”Circulation Research, vol. 94, no. 3, pp. 385–393, 2004.

[105] H. Wei, D. Zhang, L. Liu, W. Xia, and F. Li, “Rho signalingpathway enhances proliferation of PASMCs by suppressingnuclear translocation of Smad1 in PAH,” Experimental and�erapeutic Medicine, vol. 1, pp. 71–78, 2019.

[106] C. Sieber, J. Kopf, C. Hiepen, and P. Knaus, “Recent advancesin BMP receptor signaling,” Cytokine & Growth FactorReviews, vol. 20, no. 5-6, pp. 343–355, 2009.

[107] R. J. Davies and N. W. Morrell, “Molecular mechanisms ofpulmonary arterial hypertension: role of mutations in thebone morphogenetic protein type II receptor,” Chest,vol. 134, no. 6, pp. 1271–1277, 2008.

[108] L. Du, C. C. Sullivan, D. Chu et al., “Signaling molecules innonfamilial pulmonary hypertension,” New England Journalof Medicine, vol. 348, no. 6, pp. 500–509, 2003.

[109] H. Takahashi, N. Goto, Y. Kojima et al., “Downregulation oftype II bone morphogenetic protein receptor in hypoxicpulmonary hypertension,” American Journal of Physiology-Lung Cellular and Molecular Physiology, vol. 290, no. 3,pp. L450–L458, 2006.

[110] J. W. Lowery and M. P. D. Caestecker, “BMP signaling invascular development and disease,” Cytokine & GrowthFactor Reviews, vol. 21, no. 4, pp. 287–298, 2010.

[111] P. M. Hassoun, L. Mouthon, J. A. Barbera et al., “Inflam-mation, growth factors, and pulmonary vascular remodel-ing,” Journal of the American College of Cardiology, vol. 54,no. 1, pp. S10–S19, 2009.

[112] M. Rabinovitch, C. Guignabert, M. Humbert, andM. R. Nicolls, “Inflammation and immunity in the patho-genesis of pulmonary arterial hypertension,” CirculationResearch, vol. 115, no. 1, pp. 165–175, 2014.

[113] S. Shalapour and M. Karin, “Immunity, inflammation, andcancer: an eternal fight between good and evil,” Journal ofClinical Investigation, vol. 125, no. 9, pp. 3347–3355, 2015.

[114] M. Humbert, G. Monti, F. Brenot et al., “Increased inter-leukin-1 and interleukin-6 serum concentrations in severeprimary pulmonary hypertension,” American Journal ofRespiratory and Critical Care Medicine, vol. 151, no. 5,pp. 1628–1631, 1995.

[115] N. Selimovic, C.-H. Bergh, B. Andersson, E. Sakiniene,H. Carlsten, and B. Rundqvist, “Growth factors and

interleukin-6 across the lung circulation in pulmonary hy-pertension,” European Respiratory Journal, vol. 34, no. 3,pp. 662–668, 2009.

[116] L. Savale, L. Tu, D. Rideau et al., “Impact of interleukin-6 onhypoxia-induced pulmonary hypertension and lung in-flammation in mice,” Respiratory Research, vol. 10, no. 1,2009.

[117] M. K. Steiner, O. L. Syrkina, N. Kolliputi, E. J. Mark,C. A. Hales, and A. B. Waxman, “Interleukin-6 over-expression induces pulmonary hypertension,” CirculationResearch, vol. 104, no. 2, pp. 236–244, 2009.

[118] R. Paulin, A. Courboulin, J. Meloche et al., “Signal trans-ducers and activators of transcription-3/pim1 axis plays acritical role in the pathogenesis of human pulmonary arterialhypertension,” Circulation, vol. 123, no. 11, pp. 1205–1215,2011.

[119] S. Bonnet, A. Belus, J.-M. Hyvelin, E. Roux, R. Marthan, andJ.-P. Savineau, “Effect of chronic hypoxia on agonist-inducedtone and calcium signaling in rat pulmonary artery,”American Journal of Physiology-Lung Cellular and MolecularPhysiology, vol. 281, no. 1, pp. L193–L201, 2001.

[120] S. Bonnet, S. L. Archer, J. Allalunis-Turner et al., “A mito-chondria-K+ channel axis is suppressed in cancer and itsnormalization promotes apoptosis and inhibits cancergrowth,” Cancer Cell, vol. 11, no. 1, pp. 37–51, 2007.

[121] R. Savai, H. M. Al-Tamari, D. Sedding et al., “Pro-prolif-erative and inflammatory signaling converge on FoxO1transcription factor in pulmonary hypertension,” NatureMedicine, vol. 20, no. 11, pp. 1289–1300, 2014.

[122] T. B. Dansen and B. M. T. Burgering, “Unravelling the tu-mor-suppressive functions of FOXO proteins,” Trends in CellBiology, vol. 18, no. 9, pp. 421–429, 2008.

[123] M. Laplante and D. M. Sabatini, “mTOR signaling in growthcontrol and disease,” Cell, vol. 149, no. 2, pp. 274–293, 2012.

[124] E. A. Goncharova, “mTOR and vascular remodeling in lungdiseases: current challenges and therapeutic prospects,” �eFASEB Journal, vol. 27, no. 5, pp. 1796–1807, 2013.

[125] D. A. Goncharov, T. V. Kudryashova, H. Ziai et al.,“Mammalian target of rapamycin complex 2 (mTORC2)coordinates pulmonary artery smooth muscle cell meta-bolism, proliferation, and survival in pulmonary arterialhypertension,” Circulation, vol. 129, no. 8, pp. 864–874, 2014.

[126] H. Tang, K. Wu, J. Wang et al., “Pathogenic role of mTORC1and mTORC2 in pulmonary hypertension,” JACC: Basic toTranslational Science, vol. 3, no. 6, pp. 744–762, 2018.

[127] G. Halder and R. L. Johnson, “Hippo signaling: growth controland beyond,” Development, vol. 138, no. 1, pp. 9–22, 2011.

[128] U. Ehmer and J. Sage, “Control of proliferation and cancergrowth by the Hippo signaling pathway,” Molecular CancerResearch, vol. 14, no. 2, pp. 127–140, 2016.

[129] F. Zanconato, M. Forcato, G. Battilana et al., “Genome-wideassociation between YAP/TAZ/TEAD and AP-1 at en-hancers drives oncogenic growth,” Nature Cell Biology,vol. 17, no. 9, pp. 1218–1227, 2015.

[130] T. Bertero, W. M. Oldham, K. A. Cottrill et al., “Vascularstiffness mechanoactivates YAP/TAZ-dependent gluta-minolysis to drive pulmonary hypertension,” Journal ofClinical Investigation, vol. 126, no. 9, pp. 3313–3335, 2016.

[131] T. Bertero, K. A. Cottrill, Y. Lu et al., “Matrix remodelingpromotes pulmonary hypertension through feedbackmechanoactivation of the YAP/TAZ-miR-130/301 circuit,”Cell Reports, vol. 13, no. 5, pp. 1016–1032, 2015.

[132] T. V. Kudryashova, D. A. Goncharov, A. Pena et al.,“HIPPO-Integrin-linked kinase cross-talk controls self-

International Journal of Hypertension 9

sustaining proliferation and survival in pulmonary hyper-tension,” American Journal of Respiratory and Critical CareMedicine, vol. 194, no. 7, pp. 866–877, 2016.

[133] O. Boucherat, S. Bonnet, and R. Paulin, “+e HIPPO-thesisof pulmonary HYPERtension,” American Journal of Respi-ratory and Critical Care Medicine, vol. 194, no. 7, pp. 787–789, 2016.

[134] G. Kroemer and J. Pouyssegur, “Tumor cell metabolism:cancer’s Achilles’ heel,” Cancer Cell, vol. 13, no. 6,pp. 472–482, 2008.

[135] G. Sutendra and E. D. Michelakis, “+e metabolic basis ofpulmonary arterial hypertension,” Cell Metabolism, vol. 19,no. 4, pp. 558–573, 2014.

[136] M. Li, S. Riddle, H. Zhang et al., “Metabolic reprogrammingregulates the proliferative and inflammatory phenotype ofadventitial fibroblasts in pulmonary hypertension throughthe transcriptional corepressor C-terminal binding protein-1,” Circulation, vol. 134, no. 15, pp. 1105–1121, 2016.

[137] L. Plecita-Hlavata, J. Tauber, M. Li et al., “Constitutivereprogramming of fibroblast mitochondrial metabolism inpulmonary hypertension,” American Journal of RespiratoryCell and Molecular Biology, vol. 55, no. 1, pp. 47–57, 2016.

[138] P. Dromparis, R. Paulin, G. Sutendra, A. C. Qi, S. Bonnet,and E. D. Michelakis, “Uncoupling protein 2 deficiencymimics the effects of hypoxia and endoplasmic reticulumstress on mitochondria and triggers pseudohypoxic pul-monary vascular remodeling and pulmonary hypertension,”Circulation Research, vol. 113, no. 2, pp. 126–136, 2013.

[139] G. Sutendra, P. Dromparis, P. Wright et al., “+e role ofNogo and the mitochondria-endoplasmic reticulum unit inpulmonary hypertension,” Science Translational Medicine,vol. 3, no. 88, pp. 55r–88r, 2011.

[140] A.-B. Al-Mehdi, V. M. Pastukh, B. M. Swiger et al., “Peri-nuclear mitochondrial clustering creates an oxidant-richnuclear domain required for hypoxia-induced transcrip-tion,” Science Signaling, vol. 5, no. 231, p. a47, 2012.

[141] M. G. V. Heiden, L. C. Cantley, and C. B. +ompson,“Understanding the Warburg effect: the metabolic require-ments of cell proliferation,” Science, vol. 324, no. 5930,pp. 1029–1033, 2009.

[142] J. Dai, Q. Zhou, J. Chen, M. L. Rexius-Hall, J. Rehman, andG. Zhou, “Alpha-enolase regulates the malignant phenotypeof pulmonary artery smooth muscle cells via the AMPK-Aktpathway,” Nature Communications, vol. 9, no. 1, p. 3850,2018.

[143] V. Parra, R. Bravo-Sagua, I. Norambuena-Soto et al., “In-hibition of mitochondrial fission prevents hypoxia-inducedmetabolic shift and cellular proliferation of pulmonary ar-terial smooth muscle cells,” Biochimica et Biophysica Acta(BBA)-Molecular Basis of Diseas, vol. 1863, no. 11,pp. 2891–2903, 2017.

[144] L. Kovacs, Y. Cao, W. Han et al., “PFKFB3 in smooth musclepromotes vascular remodeling in pulmonary arterial hy-pertension,” American Journal of Respiratory and CriticalCare Medicine, vol. 200, no. 5, pp. 617–627, 2019.

[145] S. M. Yeligar, B.-Y. Kang, K. M. Bijli et al., “PPARgammaregulates mitochondrial structure and function and humanpulmonary artery smooth muscle cell proliferation,”American Journal of Respiratory Cell and Molecular Biology,vol. 58, no. 5, pp. 648–657, 2018.

[146] X. Xie, S. Li, Y. Zhu et al., “Egr-1 mediates leptin-inducedPPARgamma reduction and proliferation of pulmonaryartery smooth muscle cells,” Molecular Biology of the Cell,vol. 29, no. 3, pp. 356–362, 2018.

[147] H.-H. Li, H.-H. Hsu, G.-J. Chang et al., “Prostanoid EP4agonist L-902, 688 activates PPARgamma and attenuatespulmonary arterial hypertension,” American Journal ofPhysiology-Lung Cellular and Molecular Physiology, vol. 314,no. 3, pp. L349–L359, 2018.

[148] A. Weiss, M. C. Neubauer, D. Yerabolu et al., “Targetingcyclin-dependent kinases for the treatment of pulmonaryarterial hypertension,” Nature Communications, vol. 10,no. 1, p. 2204, 2019.

[149] J. Wang, W. Feng, F. Li et al., “SphK1/S1P mediates TGF-β1-induced proliferation of pulmonary artery smooth musclecells and its potential mechanisms,” Pulmonary Circulation,vol. 9, no. 1, Article ID 204589401881697, 2019.

[150] J. R. Sysol, J. Chen, S. Singla et al., “Micro-RNA-1 is de-creased by hypoxia and contributes to the development ofpulmonary vascular remodeling via regulation of sphingo-sine kinase 1,” American Journal of Physiology-Lung Cellularand Molecular Physiology, vol. 314, no. 3, pp. L461–L472,2018.

[151] L. Luo, W. Zheng, G. Lian et al., “Combination treatment ofadipose-derived stem cells and adiponectin attenuates pul-monary arterial hypertension in rats by inhibiting pulmo-nary arterial smooth muscle cell proliferation and regulatingthe AMPK/BMP/Smad pathway,” International Journal ofMolecular Medicine, vol. 1, pp. 51–60, 2017.

[152] J. Xia, L. Yang, L. Dong et al., “Cefminox, a dual agonist ofprostacyclin receptor and peroxisome proliferator-activatedreceptor-gamma identified by virtual screening, has thera-peutic efficacy against hypoxia-induced pulmonary hyper-tension in rats,” Frontiers in Pharmacology, vol. 9, p. 134,2018.

10 International Journal of Hypertension